1
|
Cabanillas B, Murdaca G, Guemari A, Torres MJ, Azkur AK, Aksoy E, Vitte J, Fernández-Santamaria R, Karavelia A, Castagnoli R, Valdelvira R, Orsi A, Ogliastro M, Massaro E, Yücel EÖ, Novak N, Agache I, Akdis M, Akdis CA. Monkeypox 2024 outbreak: Fifty essential questions and answers. Allergy 2024; 79:3285-3309. [PMID: 39495103 DOI: 10.1111/all.16374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/05/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024]
Abstract
As the world still vividly recalls the previous monkeypox (mpox) outbreak that impacted over 120 countries worldwide with more than 99,000 cases in 2022, we are now facing a second wave of infections from the monkeypox virus (MPXV), characterized by an exponential increase in cases. The current 2024 outbreak has already recorded more than 20,000 cases in Africa, marking a dramatic escalation compared to previous outbreaks. The predominance of the newly identified clade Ib variant, first detected in the Democratic Republic of the Congo (DRC) and now identified across multiple African nations and beyond, underscores its enhanced transmissibility and potential for international spread, evidenced by cases in Sweden and Thailand. The World Health Organization (WHO) declared on August 14, 2024, the current mpox outbreak a Public Health Emergency of International Concern (PHEIC), calling for heightened global public health measures. The ongoing pattern of unusual, frequent, and extensive outbreaks of mpox with potential global implications poses significant questions. This review addresses, in the format of 50 questions and answers, the 2024 mpox outbreak, detailing its characteristics, epidemiological data, and impact compared to previous outbreaks. It comprehensively explores critical questions related to MPXV virological characteristics, immunological response, clinical manifestations, epidemiology, diagnostics, and available treatments. The review also documents the significant and evolving challenges posed by the current mpox outbreak, highlighting its scale, spread, and public health response.
Collapse
Affiliation(s)
- Beatriz Cabanillas
- Department of Allergy, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Giuseppe Murdaca
- Department of Internal Medicine, University of Genova, Genova, Italy
- Allergology and Clinical Immunology Unit, San Bartolomeo Hospital, Sarzana, Italy
| | - Amir Guemari
- IDESP and PREMEDICAL, University of Montpellier-INSERM, INRIA, Montpellier, France
| | - Maria Jose Torres
- Allergy Unit, Hospital Regional Universitario de Málaga, Allergy Research Group, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, ARADyAL, Malaga University, Málaga, Spain
| | - Ahmet Kursat Azkur
- Department of Virology, Faculty of Veterinary Medicine, Kirikkale University, Kirikkale, Turkey
| | - Emel Aksoy
- Department of Virology, Faculty of Veterinary Medicine, Kirikkale University, Kirikkale, Turkey
| | - Joana Vitte
- IDESP and PREMEDICAL, University of Montpellier-INSERM, INRIA, Montpellier, France
- University of Reims Champagne-Ardenne, INSERM UMR 1250 and Immunology Laboratory, University Hospital of Reims, Reims, France
| | - Ruben Fernández-Santamaria
- Immunology Department, IIS-Fundacion Jimenez Diaz, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Aspasia Karavelia
- Department of Ear-Nose-Throat Surgery, General Hospital of Nafplio, Nafplio, Greece
| | - Riccardo Castagnoli
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Rafael Valdelvira
- Department of Allergy, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Andrea Orsi
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
- Hygiene Unit, San Martino Policlinico Hospital-IRCCS for Oncology and Neurosciences, Genova, Italy
| | - Matilde Ogliastro
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
| | - Elvira Massaro
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
| | - Esra Özek Yücel
- Division of Pediatrics, Department of Pediatric Allergy and Immunology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Natalija Novak
- Department of Dermatology and Allergy, Venusberg Campus 1, Bonn, Germany
| | - Ioana Agache
- Transylvania University, Brasov, Romania
- Theramed Medical Center, Brasov, Romania
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
2
|
Parapoxvirus Interleukin-10 Homologues Vary in Their Receptor Binding, Anti-Inflammatory, and Stimulatory Activities. Pathogens 2022; 11:pathogens11050507. [PMID: 35631028 PMCID: PMC9143231 DOI: 10.3390/pathogens11050507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 01/11/2023] Open
Abstract
Homologues of interleukin (IL)-10, a pleiotropic immunomodulatory cytokine, have been identified in the Parapoxvirus genus. The first identified, Orf virus (ORFV) IL-10, greatly enhanced infection of its host, exhibiting immune modulatory effects equivalent to human IL-10. IL-10-like genes were then identified in Bovine papular stomatitis virus (BPSV), Pseudocowpox virus (PCPV), Red deerpox virus (RDPV) and Grey sealpox virus (GSPV). This study aimed to produce and characterise recombinant parapoxvirus IL-10s, then quantitatively compare their receptor binding and immunomodulatory activities. Recombinant IL-10s were expressed, purified, then characterised using bioinformatic, biochemical and enzymatic analyses. Anti-inflammatory effects were assessed in lipoteichoic acid-activated THP-1 monocytes, and stimulatory effects in MC/9 mast cells. IL-10 receptor (IL-10R)1 binding was detected in a competitive displacement assay. BPSV IL-10 inhibited production of monocyte chemoattractant protein (MCP)-1, IL-8 and IL-1β, induced mast cell proliferation, and bound IL-10R1 similarly to ORFV IL-10. PCPV IL-10 showed reduced MCP-1 inhibition, mast cell proliferation, and IL-10R1 binding. RDPV IL-10 displayed reduced inhibition of IL-8 and MCP-1 production. GSPV IL-10 showed limited inhibition of IL-1β production and stimulation of mast cell proliferation. These findings provide valuable insight into IL-10 receptor interactions, and suggest that the parapoxvirus IL-10s play similar pathogenic roles during infection of their hosts.
Collapse
|
3
|
AlDaif BA, Mercer AA, Fleming SB. The parapoxvirus Orf virus ORF116 gene encodes an antagonist of the interferon response. J Gen Virol 2021; 102. [PMID: 34890310 DOI: 10.1099/jgv.0.001695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Orf virus (ORFV) is the type species of the Parapoxvirus genus of the Poxviridae family. Genetic and functional studies have revealed ORFV has multiple immunomodulatory genes that manipulate innate immune responses, during the early stage of infection. ORF116 is a novel gene of ORFV with hitherto unknown function. Characterization of an ORF116 deletion mutant showed that it replicated in primary lamb testis cells with reduced levels compared to the wild-type and produced a smaller plaque phenotype. ORF116 was shown to be expressed prior to DNA replication. The potential function of ORF116 was investigated by gene-expression microarray analysis in HeLa cells infected with wild-type ORFV or the ORF116 deletion mutant. The analysis of differential cellular gene expression revealed a number of interferon-stimulated genes (ISGs) differentially expressed at either 4 or 6 h post infection. IFI44 showed the greatest differential expression (4.17-fold) between wild-type and knockout virus. Other ISGs that were upregulated in the knockout included RIG-I, IFIT2, MDA5, OAS1, OASL, DDX60, ISG20 and IFIT1 and in addition the inflammatory cytokine IL-8. These findings were validated by infecting HeLa cells with an ORF116 revertant recombinant virus and analysis of transcript expression by quantitative real time-PCR (qRT-PCR). These observations suggested a role for the ORFV gene ORF116 in modulating the IFN response and inflammatory cytokines. This study represents the first functional analysis of ORF116.
Collapse
Affiliation(s)
- Basheer A AlDaif
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Andrew A Mercer
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Stephen B Fleming
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
4
|
Joshi LR, Knudsen D, Piñeyro P, Dhakal S, Renukaradhya GJ, Diel DG. Protective Efficacy of an Orf Virus-Vector Encoding the Hemagglutinin and the Nucleoprotein of Influenza A Virus in Swine. Front Immunol 2021; 12:747574. [PMID: 34804030 PMCID: PMC8602839 DOI: 10.3389/fimmu.2021.747574] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/30/2021] [Indexed: 01/19/2023] Open
Abstract
Swine influenza is a highly contagious respiratory disease of pigs caused by influenza A viruses (IAV-S). IAV-S causes significant economic losses to the swine industry and poses challenges to public health given its zoonotic potential. Thus effective IAV-S vaccines are needed and highly desirable and would benefit both animal and human health. Here, we developed two recombinant orf viruses, expressing the hemagglutinin (HA) gene (OV-HA) or the HA and the nucleoprotein (NP) genes of IAV-S (OV-HA-NP). The immunogenicity and protective efficacy of these two recombinant viruses were evaluated in pigs. Both OV-HA and OV-HA-NP recombinants elicited robust virus neutralizing antibody response in pigs, with higher levels of neutralizing antibodies (NA) being detected in OV-HA-NP-immunized animals pre-challenge infection. Although both recombinant viruses elicited IAV-S-specific T-cell responses, the frequency of IAV-S-specific proliferating CD8+ T cells upon re-stimulation was higher in OV-HA-NP-immunized animals than in the OV-HA group. Importantly, IgG1/IgG2 isotype ELISAs revealed that immunization with OV-HA induced Th2-biased immune responses, whereas immunization with OV-HA-NP virus resulted in a Th1-biased immune response. While pigs immunized with either OV-HA or OV-HA-NP were protected when compared to non-immunized controls, immunization with OV-HA-NP resulted in incremental protection against challenge infection as evidenced by a reduced secondary antibody response (NA and HI antibodies) following IAV-S challenge and reduced virus shedding in nasal secretions (lower viral RNA loads and frequency of animals shedding viral RNA and infectious virus), when compared to animals in the OV-HA group. Interestingly, broader cross neutralization activity was also observed in serum of OV-HA-NP-immunized animals against a panel of contemporary IAV-S isolates representing the major genetic clades circulating in swine. This study demonstrates the potential of ORFV-based vector for control of swine influenza virus in swine.
Collapse
Affiliation(s)
- Lok R Joshi
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States.,Department of Veterinary and Biomedical Sciences, Animal Disease Research And Diagnostic Laboratory, South Dakota State University, Brookings, SD, United States
| | - David Knudsen
- Department of Veterinary and Biomedical Sciences, Animal Disease Research And Diagnostic Laboratory, South Dakota State University, Brookings, SD, United States
| | - Pablo Piñeyro
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA, United States
| | - Santosh Dhakal
- Department of Veterinary Preventive Medicine, Center for Food Animal Health, Ohio State University, Wooster, OH, United States
| | - Gourapura J Renukaradhya
- Department of Veterinary Preventive Medicine, Center for Food Animal Health, Ohio State University, Wooster, OH, United States
| | - Diego G Diel
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States.,Department of Veterinary and Biomedical Sciences, Animal Disease Research And Diagnostic Laboratory, South Dakota State University, Brookings, SD, United States
| |
Collapse
|
5
|
Bukar AM, Jesse FFA, Abdullah CAC, Noordin MM, Lawan Z, Mangga HK, Balakrishnan KN, Azmi MLM. Immunomodulatory Strategies for Parapoxvirus: Current Status and Future Approaches for the Development of Vaccines against Orf Virus Infection. Vaccines (Basel) 2021; 9:1341. [PMID: 34835272 PMCID: PMC8624149 DOI: 10.3390/vaccines9111341] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 11/17/2022] Open
Abstract
Orf virus (ORFV), the prototype species of the parapoxvirus genus, is the causative agent of contagious ecthyma, an extremely devastating skin disease of sheep, goats, and humans that causes enormous economic losses in livestock production. ORFV is known for its ability to repeatedly infect both previously infected and vaccinated sheep due to several immunomodulatory genes encoded by the virus that temporarily suppress host immunity. Therefore, the development of novel, safe and effective vaccines against ORFV infection is an important priority. Although, the commercially licensed live-attenuated vaccines have provided partial protection against ORFV infections, the attenuated viruses have been associated with major safety concerns. In addition to safety issues, the persistent reinfection of vaccinated animals warrants the need to investigate several factors that may affect vaccine efficacy. Perhaps, the reason for the failure of the vaccine is due to the long-term adaptation of the virus in tissue culture. In recent years, the development of vaccines against ORFV infection has achieved great success due to technological advances in recombinant DNA technologies, which have opened a pathway for the development of vaccine candidates that elicit robust immunity. In this review, we present current knowledge on immune responses elicited by ORFV, with particular attention to the effects of the viral immunomodulators on the host immune system. We also discuss the implications of strain variation for the development of rational vaccines. Finally, the review will also aim to demonstrate future strategies for the development of safe and efficient vaccines against ORFV infections.
Collapse
Affiliation(s)
- Alhaji Modu Bukar
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
- Department of Science Laboratory Technology, School Agriculture and Applied Sciences, Ramat Polytechnic Maiduguri, Maiduguri 1070, Borno, Nigeria
| | - Faez Firdaus Abdullah Jesse
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | | | - Mustapha M. Noordin
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
| | - Zaharaddeen Lawan
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
| | - Hassana Kyari Mangga
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
| | - Krishnan Nair Balakrishnan
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
| | - Mohd-Lila Mohd Azmi
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
| |
Collapse
|
6
|
Costa H, Klein J, Breines EM, Nollens HH, Matassa K, Garron M, Duignan PJ, Schmitt T, Goldstein T, Tryland M. A Comparison of Parapoxviruses in North American Pinnipeds. Front Vet Sci 2021; 8:653094. [PMID: 34079832 PMCID: PMC8165162 DOI: 10.3389/fvets.2021.653094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/20/2021] [Indexed: 11/13/2022] Open
Abstract
Parapoxviruses cause nodular lesions on the skin and mucosal membranes of pinnipeds and infections by these viruses have been documented worldwide. Seal parapoxvirus is currently classified as a tentative species of the Parapoxvirus genus. Tissue or swab samples were analyzed from 11 pinnipeds of different host species undergoing rehabilitation on the east and west coasts of the United States of America (USA) that were positive for parapoxvirus. The aim of the study was to compare parapoxvirus sequences of fragments of the B2L, DNA polymerase, GIF and viral interleukin-10 ortholog (vIL-10) genes and to examine the evolutionary relationship between viruses detected in different pinniped species and at different locations with other members of the Parapoxvirus genus, such as Orf virus (ORFV), Bovine papular stomatitis virus (BPSV) and Pseudocowpox virus (PCPV). The sequence analysis showed that the parapoxvirus sequences from the pinnipeds differed significantly from those found in terrestrial hosts and that they formed a separate cluster within the genus. Our results suggest that transmission of the same parapoxvirus strain is possible between different species, including between members of different families (phocids and otariids). Animals belonging to the same species but living in distant geographic locations presented genetically distant parapoxviruses. The findings of this study demonstrate that sealpox lesions in pinnipeds of different species are caused by viruses that belong to the Parapoxvirus genus but have significant genetic differences compared to the established virus species in terrestrial hosts, thus strongly supporting the classification of pinniped parapoxvirus as a new species of the genus.
Collapse
Affiliation(s)
- Helena Costa
- Department of Arctic and Marine Biology, UiT The Arctic University of Norway, Tromsø, Norway
- Faculty of Veterinary Medicine of University of Lisbon, Lisbon, Portugal
| | - Jörn Klein
- Faculty of Health and Social Sciences, University of South-Eastern Norway, Notodden, Norway
| | - Eva M. Breines
- Department of Arctic and Marine Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | | | - Keith Matassa
- Ocean Animal Response and Research Alliance, Dana Point, CA, United States
| | - Mendy Garron
- National Oceanic and Atmospheric Administration (NOAA), Greater Atlantic Regional Fisheries Office, Gloucester, MA, United States
| | | | - Todd Schmitt
- SeaWorld Parks and Entertainment, San Diego, CA, United States
| | - Tracey Goldstein
- One Health Institute and Karen C. Drayer Wildlife Health Center, University of California, Davis, Davis, CA, United States
| | - Morten Tryland
- Department of Arctic and Marine Biology, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
7
|
Martins M, Rodrigues FS, Joshi LR, Jardim JC, Flores MM, Weiblen R, Flores EF, Diel DG. Orf virus ORFV112, ORFV117 and ORFV127 contribute to ORFV IA82 virulence in sheep. Vet Microbiol 2021; 257:109066. [PMID: 33866062 DOI: 10.1016/j.vetmic.2021.109066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/06/2021] [Indexed: 11/17/2022]
Abstract
The parapoxvirus orf virus (ORFV) encodes several immunomodulatory proteins (IMPs) that modulate host innate and pro-inflammatory responses to infection. Using the ORFV IA82 strain as the parental virus, recombinant viruses with individual deletions in the genes encoding the IMPs chemokine binding protein (CBP; ORFV112), inhibitor of granulocyte-monocyte colony-stimulating factor and IL-2 (GIF, ORFV117) and interleukin 10 homologue (vIL-10; ORFV127) were generated and characterized in vitro and in vivo. The replication properties of the individual gene deletion viruses in cell culture was not affected comparing with the parental virus. To investigate the effect of the individual gene deletions in ORFV infection and pathogenesis, groups of four lambs were inoculated with each virus and were monitored thereafter. Lambs inoculated with either recombinant or with the parental ORFV developed characteristic lesions of contagious ecthyma. The onset, nature and severity of the lesions in the oral commissure were similar in all inoculated groups from the onset (3 days post-inoculation [pi]) to the peak of clinical lesions (days 11-13 pi). Nonetheless, from days 11-13 pi onwards, the oral lesions in lambs inoculated with the recombinant viruses regressed faster than the lesions produced by the parental virus. Similarly, the amount of virus shed in the lesions were equivalent among lambs of all groups up to day 15 pi, yet they were significantly higher in the parental virus group from day 16-21 pi. In conclusion, individual deletion of these IMP genes from the ORFV genome resulted in slight reduction in virulence in vivo, as evidenced by a reduction in the duration of the clinical disease and virus shedding.
Collapse
Affiliation(s)
- Mathias Martins
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, United States; Setor de Virologia, Departamento de Medicina Veterinária Preventiva, Universidade Federal de Santa Maria, Av. Roraima 1000, prédio 63A, Santa Maria, Rio Grande do Sul, 97105-900, Brazil; Laboratório de Virologia, Medicina Veterinária, Programa de Pós-Graduação em Sanidade e Produção Animal, Universidade do Oeste de Santa Catarina, Campus II, Rodovia Rovilho Bortoluzzi, SC 480, Km 3.5, Xanxere, Santa Catarina, 89820-000, Brazil; Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, 240 Farrier Rd, Ithaca, NY, 14853, United States; Laboratório de Patologia Veterinária, Departamento de Patologia, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, Rio Grande do Sul, 97105-900, Brazil
| | - Fernando S Rodrigues
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, United States; Laboratório de Patologia Veterinária, Departamento de Patologia, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, Rio Grande do Sul, 97105-900, Brazil
| | - Lok R Joshi
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, United States; Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, 240 Farrier Rd, Ithaca, NY, 14853, United States; Laboratório de Patologia Veterinária, Departamento de Patologia, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, Rio Grande do Sul, 97105-900, Brazil
| | - José C Jardim
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, United States; Laboratório de Patologia Veterinária, Departamento de Patologia, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, Rio Grande do Sul, 97105-900, Brazil
| | - Mariana M Flores
- Setor de Virologia, Departamento de Medicina Veterinária Preventiva, Universidade Federal de Santa Maria, Av. Roraima 1000, prédio 63A, Santa Maria, Rio Grande do Sul, 97105-900, Brazil; Laboratório de Patologia Veterinária, Departamento de Patologia, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, Rio Grande do Sul, 97105-900, Brazil
| | - Rudi Weiblen
- Setor de Virologia, Departamento de Medicina Veterinária Preventiva, Universidade Federal de Santa Maria, Av. Roraima 1000, prédio 63A, Santa Maria, Rio Grande do Sul, 97105-900, Brazil; Laboratório de Patologia Veterinária, Departamento de Patologia, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, Rio Grande do Sul, 97105-900, Brazil
| | - Eduardo F Flores
- Setor de Virologia, Departamento de Medicina Veterinária Preventiva, Universidade Federal de Santa Maria, Av. Roraima 1000, prédio 63A, Santa Maria, Rio Grande do Sul, 97105-900, Brazil; Laboratório de Patologia Veterinária, Departamento de Patologia, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, Rio Grande do Sul, 97105-900, Brazil.
| | - Diego G Diel
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, United States; Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, 240 Farrier Rd, Ithaca, NY, 14853, United States; Laboratório de Patologia Veterinária, Departamento de Patologia, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, Rio Grande do Sul, 97105-900, Brazil.
| |
Collapse
|
8
|
Riad S, Xiang Y, AlDaif B, Mercer AA, Fleming SB. Rescue of a Vaccinia Virus Mutant Lacking IFN Resistance Genes K1L and C7L by the Parapoxvirus Orf Virus. Front Microbiol 2020; 11:1797. [PMID: 32903701 PMCID: PMC7438785 DOI: 10.3389/fmicb.2020.01797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 07/09/2020] [Indexed: 12/17/2022] Open
Abstract
Type 1 interferons induce the upregulation of hundreds of interferon-stimulated genes (ISGs) that combat viral replication. The parapoxvirus orf virus (ORFV) induces acute pustular skin lesions in sheep and goats and can reinfect its host, however, little is known of its ability to resist IFN. Vaccinia virus (VACV) encodes a number of factors that modulate the IFN response including the host-range genes C7L and K1L. A recombinant VACV-Western Reserve (WR) strain in which the K1L and C7L genes have been deleted does not replicate in cells treated with IFN-β nor in HeLa cells in which the IFN response is constitutive and is inhibited at the level of intermediate gene expression. Furthermore C7L is conserved in almost all poxviruses. We provide evidence that shows that although ORFV is more sensitive to IFN-β compared with VACV, and lacks homologues of KIL and C7L, it nevertheless has the ability to rescue a VACV KIL- C7L- gfp+ mutant in which gfp is expressed from a late promoter. Co-infection of HeLa cells with the mutant and ORFV demonstrated that ORFV was able to overcome the block in translation of intermediate transcripts in the mutant virus, allowing it to progress to late gene expression and new viral particles. Our findings strongly suggest that ORFV encodes a factor(s) that, although different in structure to C7L or KIL, targets an anti-viral cellular mechanism that is a highly potent at killing poxviruses.
Collapse
Affiliation(s)
- Sherief Riad
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Yan Xiang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Basheer AlDaif
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Andrew A Mercer
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Stephen B Fleming
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
9
|
Yaron JR, Zhang L, Guo Q, Burgin M, Schutz LN, Awo E, Wise L, Krause KL, Ildefonso CJ, Kwiecien JM, Juby M, Rahman MM, Chen H, Moyer RW, Alcami A, McFadden G, Lucas AR. Deriving Immune Modulating Drugs from Viruses-A New Class of Biologics. J Clin Med 2020; 9:E972. [PMID: 32244484 PMCID: PMC7230489 DOI: 10.3390/jcm9040972] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/19/2020] [Accepted: 03/23/2020] [Indexed: 02/07/2023] Open
Abstract
Viruses are widely used as a platform for the production of therapeutics. Vaccines containing live, dead and components of viruses, gene therapy vectors and oncolytic viruses are key examples of clinically-approved therapeutic uses for viruses. Despite this, the use of virus-derived proteins as natural sources for immune modulators remains in the early stages of development. Viruses have evolved complex, highly effective approaches for immune evasion. Originally developed for protection against host immune responses, viral immune-modulating proteins are extraordinarily potent, often functioning at picomolar concentrations. These complex viral intracellular parasites have "performed the R&D", developing highly effective immune evasive strategies over millions of years. These proteins provide a new and natural source for immune-modulating therapeutics, similar in many ways to penicillin being developed from mold or streptokinase from bacteria. Virus-derived serine proteinase inhibitors (serpins), chemokine modulating proteins, complement control, inflammasome inhibition, growth factors (e.g., viral vascular endothelial growth factor) and cytokine mimics (e.g., viral interleukin 10) and/or inhibitors (e.g., tumor necrosis factor) have now been identified that target central immunological response pathways. We review here current development of virus-derived immune-modulating biologics with efficacy demonstrated in pre-clinical or clinical studies, focusing on pox and herpesviruses-derived immune-modulating therapeutics.
Collapse
Affiliation(s)
- Jordan R. Yaron
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Liqiang Zhang
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Qiuyun Guo
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Michelle Burgin
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Lauren N. Schutz
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Enkidia Awo
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Lyn Wise
- University of Otago, Dunedin 9054, New Zealand; (L.W.); (K.L.K.)
| | - Kurt L. Krause
- University of Otago, Dunedin 9054, New Zealand; (L.W.); (K.L.K.)
| | | | - Jacek M. Kwiecien
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S4L8, Canada
| | - Michael Juby
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Masmudur M. Rahman
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Hao Chen
- The Department of Tumor Surgery, Second Hospital of Lanzhou University, Lanzhou 730030, China;
| | - Richard W. Moyer
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA;
| | - Antonio Alcami
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid), Cantoblanco, 28049 Madrid, Spain;
| | - Grant McFadden
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Alexandra R. Lucas
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
- St Joseph Hospital, Dignity Health, Creighton University, Phoenix, AZ 85013, USA
| |
Collapse
|
10
|
Andreani J, Fongue J, Bou Khalil JY, David L, Mougari S, Le Bideau M, Abrahão J, Berbis P, La Scola B. Human Infection with Orf Virus and Description of Its Whole Genome, France, 2017. Emerg Infect Dis 2019; 25:2197-2204. [PMID: 31742503 PMCID: PMC6874271 DOI: 10.3201/eid2512.181513] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Zoonotic transmission of parapoxvirus from animals to humans has been reported; clinical manifestations are skin lesions on the fingers and hands after contact with infected animals. We report a human infection clinically suspected as being ecthyma contagiosum. The patient, a 65-year-old woman, had 3 nodules on her hands. She reported contact with a sheep during the Aïd-el-Fitr festival in France during 2017. We isolated the parapoxvirus orf virus from these nodules by using a nonconventional cell and sequenced the orf genome. We identified a novel orf virus genome and compared it with genomes of other orf viruses. More research is needed on the genus Parapoxvirus to understand worldwide distribution of and infection by orf virus, especially transmission between goats and sheep.
Collapse
|
11
|
Wang R, Wang Y, Liu F, Luo S. Orf virus: A promising new therapeutic agent. Rev Med Virol 2018; 29:e2013. [PMID: 30370570 DOI: 10.1002/rmv.2013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/14/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022]
Abstract
The orf virus (ORFV) is a zoonotic, epitheliotropic, DNA parapoxvirus that infects principally sheep and goats. Exposure of animals to the virus or immunization by an ORFV preparation can accentuate the severity of disease, which has provoked an interest in the underlying cellular, virological, and molecular mechanisms. The identified ORFV virulence genes and the fact that the virus can repeatedly infect a host, owing to its evasive mechanisms, contribute to the development of potent immune modulators in various animal species. ORFV has been developed as a vaccine in veterinary medicine. The unique host immune-evasion ability of ORFV has made it an important candidate for vaccine vectors and biological agents (as an oncolytic virus). Genetic modifications using ORFV to obtain safe and efficient preparations and mechanistic studies are improvements to the currently available methods for disease treatment.
Collapse
Affiliation(s)
- Ruixue Wang
- Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China.,Department of Basic Medical Sciences, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Yong Wang
- Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Fang Liu
- Department of Basic Medical Sciences, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Shuhong Luo
- Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| |
Collapse
|
12
|
Characterisation of putative immunomodulatory gene knockouts of lumpy skin disease virus in cattle towards an improved vaccine. Vaccine 2018; 36:4708-4715. [PMID: 29941325 DOI: 10.1016/j.vaccine.2018.06.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 06/05/2018] [Accepted: 06/06/2018] [Indexed: 11/22/2022]
Abstract
Lumpy skin disease virus (LSDV) is responsible for causing severe economic losses to cattle farmers throughout Africa, the Middle East, and more recently, South-Eastern Europe and Russia. It belongs to the Capripoxvirus genus of the Poxviridae family, with closely related sheeppox and goatpox viruses. Like other poxviruses, the viral genome codes for a number of genes with putative immunomodulatory capabilities. Current vaccines for protecting cattle against lumpy skin disease (LSD) based on live-attenuated strains of field isolates passaged by cell culture, resulting in random mutations. Although generally effective, these vaccines can have drawbacks, including injection site reactions and/or limited immunogenicity. A pilot study was conducted using a more targeted approach where two putative immunomodulatory genes were deleted separately from the genome of a virulent LSDV field isolate. These were open reading frame (ORF) 005 and ORF008, coding for homologues of an interleukin 10-like and interferon-gamma receptor-like gene, respectively. The resulting knockout constructs were evaluated in cattle for safety, immunogenicity and protection. Severe post-vaccinal reactions and febrile responses were observed for both constructs. Two calves inoculated with the ORF008 knockout construct developed multiple lesions and were euthanised. Following challenge, none of the animals inoculated with the knockout constructs showed any external clinical signs of LSD, compared to the negative controls. Improved cellular and humoral immune responses were recorded in both of these groups compared to the positive control. The results indicate that at the high inoculation doses used, the degree of attenuation achieved was insufficient for further use in cattle due to the adverse reactions observed.
Collapse
|
13
|
Martins M, Joshi LR, Rodrigues FS, Anziliero D, Frandoloso R, Kutish GF, Rock DL, Weiblen R, Flores EF, Diel DG. Immunogenicity of ORFV-based vectors expressing the rabies virus glycoprotein in livestock species. Virology 2017; 511:229-239. [DOI: 10.1016/j.virol.2017.08.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/16/2017] [Accepted: 08/18/2017] [Indexed: 02/06/2023]
|
14
|
Jia H, Zhan L, Wang X, He X, Chen G, Zhang Y, Feng Y, Wei Y, Zhang Y, Jing Z. Transcriptome analysis of sheep oral mucosa response to Orf virus infection. PLoS One 2017; 12:e0186681. [PMID: 29073164 PMCID: PMC5658058 DOI: 10.1371/journal.pone.0186681] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 10/05/2017] [Indexed: 01/15/2023] Open
Abstract
Contagious ecthyma is a highly contagious disease with worldwide distribution, which is caused by the Orf virus (ORFV) belonging to the Parapoxvirus. To study the alteration of host gene expression in response to ORFV infection at the transcriptional level, several young small-tailed Han sheep were inoculated with ORFV, and their oral mucosa tissue samples (T0, T3, T7 and T15) were collected on day 0, 3, 7 and 15 after ORFV infection respectively. RNA-seq transcriptome comparisons were performed, showing that 1928, 3219 and 2646 differentially expressed genes (DEGs) were identified among T3 vs. T0, T7 vs. T0, and T15 vs. T0 respectively. Gene Ontology (GO) analyses of the DEGs from these comparisons, revealed that ORFV might provoke vigorous immune response of the host cells during the early stage of infection. Moreover, GO and network analysis showed that positive and negative regulative mechanisms of apoptosis were integrated in the host cells through up or down-regulating the expression level of DEGs involved in apoptotic pathways, in order to reach a homeostasis of oral mucosa tissues during the exposure to ORFV infection. In conclusion, our study for the first time describes the direct effects of ORFV on the global host gene expression of its host using high-throughput RNA sequencing, which provides a resource for future characterizing the interaction mechanism between the mammalian host and ORFV.
Collapse
Affiliation(s)
- Huaijie Jia
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Leilei Zhan
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, China
| | - Xiaoxia Wang
- School of Public Health, Faculty of Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaobing He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Guohua Chen
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yu Zhang
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, China
| | - Yuan Feng
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yaxun Wei
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, China
| | - Yi Zhang
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, China
- Laboratory for Genome Regulation and Human Health, ABLife Inc., Wuhan, Hubei, China
| | - Zhizhong Jing
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- * E-mail:
| |
Collapse
|
15
|
Abstract
We recovered the first full-length poxvirus genome, including the terminal hairpin region, directly from complex clinical material using a combination of second generation short read and third generation nanopore sequencing technologies. The complete viral genome sequence was directly recovered from a skin lesion of a grey seal thereby preventing sequence changes due to in vitro passaging of the virus. Subsequent analysis of the proteins encoded by this virus identified genes specific for skin adaptation and pathogenesis of parapoxviruses. These data warrant the classification of seal parapoxvirus, tentatively designated SePPV, as a new species within the genus Parapoxvirus.
Collapse
|
16
|
Fleming SB, McCaughan C, Lateef Z, Dunn A, Wise LM, Real NC, Mercer AA. Deletion of the Chemokine Binding Protein Gene from the Parapoxvirus Orf Virus Reduces Virulence and Pathogenesis in Sheep. Front Microbiol 2017; 8:46. [PMID: 28174562 PMCID: PMC5258736 DOI: 10.3389/fmicb.2017.00046] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 01/06/2017] [Indexed: 12/25/2022] Open
Abstract
Orf virus (ORFV) is the type species of the Parapoxvirus genus of the family Poxviridae and infects sheep and goats, often around the mouth, resulting in acute pustular skin lesions. ORFV encodes several secreted immunomodulators including a broad-spectrum chemokine binding protein (CBP). Chemokines are a large family of secreted chemotactic proteins that activate and regulate inflammation induced leukocyte recruitment to sites of infection. In this study we investigated the role of CBP in vivo in the context of ORFV infection of sheep. The CBP gene was deleted from ORFV strain NZ7 and infections of sheep used to investigate the effect of CBP on pathogenesis. Animals were either infected with the wild type (wt) virus, CBP-knockout virus or revertant strains. Sheep were infected by scarification on the wool-less area of the hind legs at various doses of virus. The deletion of the CBP gene severely attenuated the virus, as only few papules formed when animals were infected with the CBP-knock-out virus at the highest dose (107 p.f.u). In contrast, large pustular lesions formed on almost all animals infected with the wt and revertant strains at 107 p.f.u. The lesions for the CBP-knock-out virus resolved approximately 5–6 days p.i, at a dose of 107 pfu whereas in animals infected with the wt and revertants at this dose, lesions began to resolve at approximately 10 days p.i. Few pustules developed at the lowest dose of 103 p.f.u. for all viruses. Immunohistochemistry of biopsy skin-tissue from pustules showed that the CBP-knockout virus replicated in all animals at the highest dose and was localized to the skin epithelium while haematoxylin and eosin staining showed histological features of the CBP-knockout virus typical of the parent virus with acanthosis, elongated rete ridges and orthokeratotic hyperkeratosis. MHC-II immunohistochemistry analysis for monocytes and dendritic cells showed greater staining within the papillary dermis of the CBP-knock-out virus compared with the revertant viruses, however this was not the case with the wt where staining was similar. Our results show that the CBP gene encodes a secreted immunodulator that has a critical role in virulence and pathogenesis.
Collapse
Affiliation(s)
- Stephen B Fleming
- Department of Microbiology and Immunology, University of Otago Dunedin, New Zealand
| | - Catherine McCaughan
- Department of Microbiology and Immunology, University of Otago Dunedin, New Zealand
| | - Zabeen Lateef
- Department of Microbiology and Immunology, University of Otago Dunedin, New Zealand
| | - Amy Dunn
- Department of Microbiology and Immunology, University of Otago Dunedin, New Zealand
| | - Lyn M Wise
- Department of Microbiology and Immunology, University of Otago Dunedin, New Zealand
| | - Nicola C Real
- Department of Microbiology and Immunology, University of Otago Dunedin, New Zealand
| | - Andrew A Mercer
- Department of Microbiology and Immunology, University of Otago Dunedin, New Zealand
| |
Collapse
|
17
|
Wang L, Lu B, Zheng H, Zhang K, Liu X. Parapoxvirus orf virus infection induces an increase in interleukin-8, tumour necrosis factor-α, and decorin in goat skin fibroblast cells. J Vet Res 2016. [DOI: 10.1515/jvetres-2016-0036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Abstract
Introduction: Orf virus (ORFV) is a prototype Parapoxvirus species in the Poxviridae family that causes serious zoonotic infectious disease. Goat skin fibroblast (GSF) cells are the major host targets of ORFV. Interleukin 8 (IL-8) and tumour necrosis factor (TNF)-α are known to play a vital role in immune response during viral infections. However, the manner of variation over time of their level of expression in GSF cells remains unclear.
Material and Methods: In this study, quantitative enzyme-linked immunosorbent assay chips were used to detect changes in the levels of these cytokines expressed and secreted in GSF cells after ORFV infection.
Results: Results showed that the expression of IL-8, TNF-α, and decorin was upregulated in the cell lysates, and that secreted decorin and IL-8 were significantly increased in cell supernatant.
Conclusion: The results provided possible approaches to elucidation of how ORFV infection initiates host cell immune response.
Collapse
Affiliation(s)
- Lingling Wang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Bingzhou Lu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Haixue Zheng
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Keshan Zhang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Xiangtao Liu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| |
Collapse
|
18
|
Hain KS, Joshi LR, Okda F, Nelson J, Singrey A, Lawson S, Martins M, Pillatzki A, Kutish GF, Nelson EA, Flores EF, Diel DG. Immunogenicity of a recombinant parapoxvirus expressing the spike protein of Porcine epidemic diarrhea virus. J Gen Virol 2016; 97:2719-2731. [PMID: 27558814 DOI: 10.1099/jgv.0.000586] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The parapoxvirus Orf virus (ORFV), has long been recognized for its immunomodulatory properties in permissive and non-permissive animal species. Here, a new recombinant ORFV expressing the full-length spike (S) protein of Porcine epidemic diarrhea virus (PEDV) was generated and its immunogenicity and protective efficacy were evaluated in pigs. The PEDV S was inserted into the ORFV121 gene locus, an immunomodulatory gene that inhibits activation of the NF-κB signalling pathway and contributes to ORFV virulence in the natural host. The recombinant ORFV-PEDV-S virus efficiently and stably expressed the PEDV S protein in cell culture in vitro. Three intramuscular (IM) immunizations with the recombinant ORFV-PEDV-S in 3-week-old pigs elicited robust serum IgG, IgA and neutralizing antibody responses against PEDV. Additionally, IM immunization with the recombinant ORFV-PEDV-S virus protected pigs from clinical signs of porcine epidemic diarrhoea (PED) and reduced virus shedding in faeces upon challenge infection. These results demonstrate the suitability of ORFV121 gene locus as an insertion site for heterologous gene expression and delivery by ORFV-based viral vectors. Additionally, the results provide evidence of the potential of ORFV as a vaccine delivery vector for enteric viral diseases of swine. This study may have important implications for future development of ORFV-vectored vaccines for swine.
Collapse
Affiliation(s)
- Kyle S Hain
- Animal Disease Research and Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Lok R Joshi
- Animal Disease Research and Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Faten Okda
- Animal Disease Research and Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA.,National Research Center, Giza, Egypt
| | - Julie Nelson
- Animal Disease Research and Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Aaron Singrey
- Animal Disease Research and Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Steven Lawson
- Animal Disease Research and Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Mathias Martins
- Department of Preventive Veterinary Medicine, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Angela Pillatzki
- Animal Disease Research and Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Gerald F Kutish
- Department of Pathobiology, University of Connecticut, Storrs, CT, USA
| | - Eric A Nelson
- Animal Disease Research and Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Eduardo F Flores
- Department of Preventive Veterinary Medicine, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Diego G Diel
- Animal Disease Research and Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA
| |
Collapse
|
19
|
Wang X, Zhang J, Hao W, Peng Y, Li H, Li W, Li M, Luo S. Isolation and Characterization of Monoclonal Antibodies Against a Virion Core Protein of Orf Virus Strain NA1/11 As Potential Diagnostic Tool for Orf Viruses. Monoclon Antib Immunodiagn Immunother 2016; 34:233-45. [PMID: 26301926 DOI: 10.1089/mab.2014.0101] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Orf is caused by the orf virus (ORFV) and is a non-systemic, widespread disease afflicting sheep, goats, wild ruminants, and humans. Recent outbreaks in sheep and goats in Jilin and other northern Chinese provinces raise concerns about orf control in China. Thirty-five hybridoma clones were constructed from splenocytes of BALB/c mice immunized with natural orf virus protein. These hybridomas were used to produce antibodies targeting ORFV proteins. Immunological characterization of these monoclonal antibodies (MAb) showed that the 5F2D8 hybridoma line produced MAb that can recognize the 100, 70, and 20 kDa bands from total viral lysate. This hybridoma was further characterized by immunoprecipitation and peptide sequencing. The results indicate that 5F2D8 specifically recognizes orf virus encoded protein ORFV086, a late expression virion core protein that plays important roles in progeny virus particle assembly, morphogenesis, and maturity. Further experiments demonstrate that this MAb did not react with other viral proteins of ORFV orthopoxviruses, but reacted strongly to different field isolates of orf viruses from China. Additionally, this anti-ORFV086 MAb possesses ORFV neutralizing capability. Sequence alignments and phylogenetic analysis determined that ORFV086 of NA1/11, clustered together with NZ2 and IA82, is highly conserved and has structural similarities with the Vaccinia virus core protein P4a. As such, this MAb has great potential as a diagnostic tool for orf viruses, in the further exploration of orf pathogenesis, and in disease control and prevention.
Collapse
Affiliation(s)
- Xiaoping Wang
- Institute of Antibody Engineering, School of Biotechnology, Southern Medical University , Guangzhou, People's Republic of China
| | - Jiafeng Zhang
- Institute of Antibody Engineering, School of Biotechnology, Southern Medical University , Guangzhou, People's Republic of China
| | - Wenbo Hao
- Institute of Antibody Engineering, School of Biotechnology, Southern Medical University , Guangzhou, People's Republic of China
| | - Yongzheng Peng
- Institute of Antibody Engineering, School of Biotechnology, Southern Medical University , Guangzhou, People's Republic of China
| | - Hong Li
- Institute of Antibody Engineering, School of Biotechnology, Southern Medical University , Guangzhou, People's Republic of China
| | - Wei Li
- Institute of Antibody Engineering, School of Biotechnology, Southern Medical University , Guangzhou, People's Republic of China
| | - Ming Li
- Institute of Antibody Engineering, School of Biotechnology, Southern Medical University , Guangzhou, People's Republic of China
| | - Shuhong Luo
- Institute of Antibody Engineering, School of Biotechnology, Southern Medical University , Guangzhou, People's Republic of China
| |
Collapse
|
20
|
Orf virus IL-10 reduces monocyte, dendritic cell and mast cell recruitment to inflamed skin. Virus Res 2016; 213:230-237. [DOI: 10.1016/j.virusres.2015.12.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/14/2015] [Accepted: 12/21/2015] [Indexed: 12/17/2022]
|
21
|
Boshra H, Truong T, Nfon C, Bowden TR, Gerdts V, Tikoo S, Babiuk LA, Kara P, Mather A, Wallace DB, Babiuk S. A lumpy skin disease virus deficient of an IL-10 gene homologue provides protective immunity against virulent capripoxvirus challenge in sheep and goats. Antiviral Res 2015; 123:39-49. [PMID: 26341190 DOI: 10.1016/j.antiviral.2015.08.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 08/27/2015] [Accepted: 08/31/2015] [Indexed: 10/23/2022]
Abstract
Sheep and goat pox continue to be important livestock diseases that pose a major threat to the livestock industry in many regions in Africa and Asia. Currently, several live attenuated vaccines are available and used in endemic countries to control these diseases. One of these is a partially attenuated strain of lumpy skin disease virus (LSDV), KS-1, which provides cross-protection against both sheep pox and goat pox. However, when used in highly stressed dairy cattle to protect against lumpy skin disease (LSD) the vaccine can cause clinical disease. In order to develop safer vaccines effective against all three diseases, a pathogenic strain of LSDV (Warmbaths [WB], South Africa) was attenuated by removing a putative virulence factor gene (IL-10-like) using gene knockout (KO) technology. This construct (LSDV WB005KO) was then evaluated as a vaccine for sheep and goats against virulent capripoxvirus challenge. Sheep and goats were vaccinated with the construct and the animals were observed for 21days. The vaccine appeared to be safe, and did not cause disease, although it induced minor inflammation at the injection site similar to that caused by other attenuated sheep and goat pox vaccines. In addition, no virus replication was detected in blood, oral or nasal swabs using real-time PCR following vaccination and low levels of neutralising antibodies were detected in both sheep and goats. Leukocytes isolated from vaccinated animals following vaccination elicited capripoxvirus-specific IFN-γ secretion, suggesting that immunity was also T-cell mediated. Following challenge with virulent capripoxvirus, vaccinated sheep and goats were found to be completely protected and exhibited no clinical disease. Furthermore, real-time PCR of blood samples at various time points suggested that viremia was absent in both groups of vaccinated animals, as opposed to capripoxvirus-related clinical disease and viremia observed in the unvaccinated animals. These findings suggest that this novel knockout strain of LSDV has potential as a vaccine to protect livestock against sheep pox and goat pox.
Collapse
Affiliation(s)
- Hani Boshra
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | - Thang Truong
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | - Charles Nfon
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | - Timothy R Bowden
- CSIRO Biosecurity Flagship, Australian Animal Health Laboratory, Geelong, Australia
| | - Volker Gerdts
- Vaccine & Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
| | - Suresh Tikoo
- Vaccine & Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada; School of Public Health, University of Saskatchewan, Saskatoon, SK, Canada
| | | | - Pravesh Kara
- ARC-Onderstepoort Veterinary Institute, Onderstepoort, South Africa
| | - Arshad Mather
- ARC-Onderstepoort Veterinary Institute, Onderstepoort, South Africa
| | - David B Wallace
- ARC-Onderstepoort Veterinary Institute, Onderstepoort, South Africa; Department Veterinary Tropical Diseases, Faculty Veterinary Science, University of Pretoria, South Africa
| | - Shawn Babiuk
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada; University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
22
|
Fleming SB, Wise LM, Mercer AA. Molecular genetic analysis of orf virus: a poxvirus that has adapted to skin. Viruses 2015; 7:1505-39. [PMID: 25807056 PMCID: PMC4379583 DOI: 10.3390/v7031505] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 03/17/2015] [Accepted: 03/19/2015] [Indexed: 12/17/2022] Open
Abstract
Orf virus is the type species of the Parapoxvirus genus of the family Poxviridae. It induces acute pustular skin lesions in sheep and goats and is transmissible to humans. The genome is G+C rich, 138 kbp and encodes 132 genes. It shares many essential genes with vaccinia virus that are required for survival but encodes a number of unique factors that allow it to replicate in the highly specific immune environment of skin. Phylogenetic analysis suggests that both viral interleukin-10 and vascular endothelial growth factor genes have been "captured" from their host during the evolution of the parapoxviruses. Genes such as a chemokine binding protein and a protein that binds granulocyte-macrophage colony-stimulating factor and interleukin-2 appear to have evolved from a common poxvirus ancestral gene while three parapoxvirus nuclear factor (NF)-κB signalling pathway inhibitors have no homology to other known NF-κB inhibitors. A homologue of an anaphase-promoting complex subunit that is believed to manipulate the cell cycle and enhance viral DNA synthesis appears to be a specific adaptation for viral-replication in keratinocytes. The review focuses on the unique genes of orf virus, discusses their evolutionary origins and their role in allowing viral-replication in the skin epidermis.
Collapse
Affiliation(s)
- Stephen B Fleming
- Department of Microbiology and Immunology, 720 Cumberland St, University of Otago, Dunedin 9016, New Zealand.
| | - Lyn M Wise
- Department of Microbiology and Immunology, 720 Cumberland St, University of Otago, Dunedin 9016, New Zealand.
| | - Andrew A Mercer
- Department of Microbiology and Immunology, 720 Cumberland St, University of Otago, Dunedin 9016, New Zealand.
| |
Collapse
|
23
|
Martins M, Cargnelutti JF, Weiblen R, Flores EF. Pathogenesis in lambs and sequence analysis of putative virulence genes of Brazilian orf virus isolates. Vet Microbiol 2014; 174:69-77. [PMID: 25293399 DOI: 10.1016/j.vetmic.2014.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 09/09/2014] [Accepted: 09/12/2014] [Indexed: 11/18/2022]
Abstract
The parapoxvirus orf virus (ORFV) is the agent of contagious ecthyma, an ubiquitous mucocutaneous disease of sheep and goats that may present variable clinical presentations. We herein studied the pathogenesis of ORFV infection in lambs and analyzed three putative virulence genes of four Brazilian ORFV isolates. Lambs inoculated in the labial commissures with each ORFV isolate (n=4, viral titer 10(5.6) TCID50/ml) developed classical orf lesions, characterized by a progressive course of erythema/macules, vesicles, pustules and proliferative scabs. Lesions lasted an average of 22.9 days (18-26) and virus shedding was detected for approximately 24.6 days (18-30). Two isolates (SV269/11 and SV820/10) produced more severe, long-lasting lesions resulting in highest clinical scores. Lambs inoculated with isolate SV581/11 developed lesions markedly milder (lower clinical scores [p<0.05]) and more limited than the other groups. Virus shedding by SV581/11 group, however, lasted similarly or even longer than the other groups. Sequence analysis of three virulence genes (VEGF, VIR and IL-10v) revealed amino acid deletions and mutations in VEGF and IL-10v genes of SV581/11 and SV252/11, the isolate(s) producing milder lesions. Additionally, the VEGF gene of isolate SV581/11 presented the lowest amino acid identity with the other isolates and with ORFV standard strain OV-IA82. Thus, these results demonstrate that ORFV isolates may display differential virulence in lambs and these differences might be associated with genetic changes in putative virulence genes.
Collapse
Affiliation(s)
- Mathias Martins
- Setor de Virologia, Department of Preventive Veterinary Medicine, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, RS 97105-900, Brazil
| | - Juliana F Cargnelutti
- Setor de Virologia, Department of Preventive Veterinary Medicine, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, RS 97105-900, Brazil
| | - Rudi Weiblen
- Setor de Virologia, Department of Preventive Veterinary Medicine, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, RS 97105-900, Brazil
| | - Eduardo F Flores
- Setor de Virologia, Department of Preventive Veterinary Medicine, Universidade Federal de Santa Maria, Av. Roraima, 1000, Santa Maria, RS 97105-900, Brazil.
| |
Collapse
|
24
|
Hosamani M, Scagliarini A, Bhanuprakash V, McInnes CJ, Singh RK. Orf: an update on current research and future perspectives. Expert Rev Anti Infect Ther 2014; 7:879-93. [PMID: 19735227 DOI: 10.1586/eri.09.64] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Madhusudan Hosamani
- Indian Veterinary Research Institute, Mukteswar-263138, Nainital Distt., India and Indian Veterinary Research Institute, Hebbal, Bangalore-24, India.
| | | | | | | | | |
Collapse
|
25
|
Eberhardt MK, Barry PA. Pathogen manipulation of cIL-10 signaling pathways: opportunities for vaccine development? Curr Top Microbiol Immunol 2014; 380:93-128. [PMID: 25004815 DOI: 10.1007/978-3-662-43492-5_5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Interleukin-10 (IL-10) is a tightly regulated, pleiotropic cytokine that has profound effects on all facets of the immune system, eliciting cell-type-specific responses within cells expressing the IL-10 receptor (IL-10R). It is considered a master immune regulator, and imbalances in IL-10 expression, resulting from either inherent or infectious etiologies, have far reaching clinical ramifications. Regarding infectious diseases, there has been accumulating recognition that many pathogens, particularly those that establish lifelong persistence, share a commonality of their natural histories: manipulation of IL-10-mediated signaling pathways. Multiple viral, bacterial, protozoal, and fungal pathogens appear to have evolved mechanisms to co-opt normal immune functions, including those involving IL-10R-mediated signaling, and immune effector pathways away from immune-mediated protection toward environments of immune evasion, suppression, and tolerance. As a result, pathogens can persist for the life of the infected host, many of whom possess otherwise competent immune systems. Because of pathogenic avoidance of immune clearance, persistent infections can exact incalculable physical and financial costs, and represent some of the most vexing challenges for improvements in human health. Enormous benefits could be gained by the development of efficient prevention and/or therapeutic strategies that block primary infection, or clear the infection. There are now precedents that indicate that modalities focusing on pathogen-mediated manipulation of IL-10 signaling may have clinical benefit.
Collapse
Affiliation(s)
- Meghan K Eberhardt
- Center for Comparative Medicine, University of California, Davis, CA, 95616, USA
| | | |
Collapse
|
26
|
Ouyang P, Rakus K, van Beurden SJ, Westphal AH, Davison AJ, Gatherer D, Vanderplasschen AF. IL-10 encoded by viruses: a remarkable example of independent acquisition of a cellular gene by viruses and its subsequent evolution in the viral genome. J Gen Virol 2013; 95:245-262. [PMID: 24225498 DOI: 10.1099/vir.0.058966-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Many viruses have evolved strategies to deregulate the host immune system. These strategies include mechanisms to subvert or recruit the host cytokine network. IL-10 is a pleiotropic cytokine that has both immunostimulatory and immunosuppressive properties. However, its key features relate mainly to its capacity to exert potent immunosuppressive effects. Several viruses have been shown to upregulate the expression of cellular IL-10 (cIL-10) with, in some cases, enhancement of infection by suppression of immune functions. Other viruses encode functional orthologues of cIL-10, called viral IL-10s (vIL-10s). The present review is devoted to these virokines. To date, vIL-10 orthologues have been reported for 12 members of the family Herpesviridae, two members of the family Alloherpesviridae and seven members of the family Poxviridae. Study of vIL-10s demonstrated several interesting aspects on the origin and the evolution of these viral genes, e.g. the existence of multiple (potentially up to nine) independent gene acquisition events at different times during evolution, viral gene acquisition resulting from recombination with cellular genomic DNA or cDNA derived from cellular mRNA and the evolution of cellular sequence in the viral genome to restrict the biological activities of the viral orthologues to those beneficial for the virus life cycle. Here, various aspects of the vIL-10s described to date are reviewed, including their genetic organization, protein structure, origin, evolution, biological properties and potential in applied research.
Collapse
Affiliation(s)
- Ping Ouyang
- Immunology-Vaccinology (B43b), Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Krzysztof Rakus
- Immunology-Vaccinology (B43b), Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Steven J van Beurden
- Immunology-Vaccinology (B43b), Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Adrie H Westphal
- Laboratory of Biochemistry, Department of Agrotechnology and Food Sciences, Wageningen University, Wageningen UR, Dreijenlaan 3, 6703 HA Wageningen, The Netherlands
| | - Andrew J Davison
- MRC-University of Glasgow Centre for Virus Research, 8 Church Street, Glasgow G11 5JR, UK
| | - Derek Gatherer
- Division of Biomedical & Life Sciences, Lancaster University, Lancaster LA1 4YQ, UK.,MRC-University of Glasgow Centre for Virus Research, 8 Church Street, Glasgow G11 5JR, UK
| | - Alain F Vanderplasschen
- Immunology-Vaccinology (B43b), Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| |
Collapse
|
27
|
Vaccination against a virus-encoded cytokine significantly restricts viral challenge. J Virol 2013; 87:11323-31. [PMID: 23946461 DOI: 10.1128/jvi.01925-13] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Identification of immune correlates of protection for viral vaccines is complicated by multiple factors, but there is general consensus on the importance of antibodies that neutralize viral attachment to susceptible cells. Development of new viral vaccines has mostly followed this neutralizing antibody paradigm, but as a recent clinical trial of human cytomegalovirus (HCMV) vaccination demonstrated, this singular approach can yield limited protective efficacy. Since HCMV devotes >50% of its coding capacity to proteins that modulate host immunity, it is hypothesized that expansion of vaccine targets to include this part of the viral proteome will disrupt viral natural history. HCMV and rhesus cytomegalovirus (RhCMV) each encode an ortholog to the cellular interleukin-10 (cIL-10) cytokine: cmvIL-10 and rhcmvIL10, respectively. Despite extensive sequence divergence from their host's cIL-10, each viral IL-10 retains nearly identical functionality to cIL-10. Uninfected rhesus macaques were immunized with engineered, nonfunctional rhcmvIL-10 variants, which were constructed by site-directed mutagenesis to abolish binding to the cIL-10 receptor. Vaccinees developed antibodies that neutralized rhcmvIL-10 function with no cross-neutralization of cIL-10. Following subcutaneous RhCMV challenge, the vaccinees exhibited both reduced RhCMV replication locally at the inoculation site and systemically and significantly reduced RhCMV shedding in bodily fluids compared to controls. Attenuation of RhCMV infection by rhcmvIL-10 vaccination argues that neutralization of viral immunomodulation may be a new vaccine paradigm for HCMV by expanding potential vaccine targets.
Collapse
|
28
|
Ouyang P, Rakus K, Boutier M, Reschner A, Leroy B, Ronsmans M, Fournier G, Scohy S, Costes B, Wattiez R, Vanderplasschen A. The IL-10 homologue encoded by cyprinid herpesvirus 3 is essential neither for viral replication in vitro nor for virulence in vivo. Vet Res 2013; 44:53. [PMID: 23865540 PMCID: PMC3750702 DOI: 10.1186/1297-9716-44-53] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 06/10/2013] [Indexed: 01/09/2023] Open
Abstract
Cyprinid herpesvirus 3 (CyHV-3), a member of the family Alloherpesviridae, is the causative agent of a lethal disease in common and koi carp. CyHV-3 ORF134 encodes an interleukin-10 (IL-10) homologue. The present study was devoted to this ORF. Transcriptomic analyses revealed that ORF134 is expressed as a spliced gene belonging to the early-late class. Proteomic analyses of CyHV-3 infected cell supernatant demonstrated that the ORF134 expression product is one of the most abundant proteins of the CyHV-3 secretome. To investigate the role of ORF134 in viral replication in vitro and in virulence in vivo, a deleted strain and a derived revertant strain were produced using BAC cloning technologies. The recombinant ORF134 deleted strain replicated in vitro comparably to the parental and the revertant strains. Infection of fish by immersion in water containing the virus induced comparable CyHV-3 disease for the three virus genotypes tested (wild type, deleted and revertant). Quantification of viral DNA by real time TaqMan PCR (in the gills and the kidney) and analysis of carp cytokine expression (in the spleen) by RT-qPCR at different times post-infection did not revealed any significant difference between the groups of fish infected with the three virus genotypes. Similarly, histological examination of the gills and the kidney of infected fish revealed no significant differences between fish infected with ORF134 deleted virus versus fish infected with the control parental or revertant strains. All together, the results of the present study demonstrate that the IL-10 homologue encoded by CyHV-3 is essential neither for viral replication in vitro nor for virulence in common carp.
Collapse
Affiliation(s)
- Ping Ouyang
- Immunology-Vaccinology (B43b), Department of Infectious and Parasitic Diseases (B43b), Faculty of Veterinary Medicine, University of Liège, Liège, B-4000, Belgium.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Nagarajan G, Swami SK, Dahiya SS, Sivakumar G, Narnaware S, Tuteja F, Patil N. Comparison of virokine from camel pseudocowpoxvirus (PCPV) with Interleukin 10 of the Dromedary camel (Camelus dromedarius). Cytokine 2013; 61:356-9. [DOI: 10.1016/j.cyto.2012.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 10/10/2012] [Accepted: 12/06/2012] [Indexed: 11/29/2022]
|
30
|
Li W, Ning Z, Hao W, Song D, Gao F, Zhao K, Liao X, Li M, Rock DL, Luo S. Isolation and phylogenetic analysis of orf virus from the sheep herd outbreak in northeast China. BMC Vet Res 2012; 8:229. [PMID: 23174032 PMCID: PMC3561078 DOI: 10.1186/1746-6148-8-229] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 10/16/2012] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Orf is a zoonotic and epitheliotrophic contagious disease that mainly affects sheep, goats, wild ruminants, and humans with a worldwide distribution. To date, there is little information on the characterization of ORFV strains that are endemic in Mainland China. In addition, the relationship between the severity of disease and the molecular profile of ORFV strains has not been fully elucidated. RESULTS From the recent outbreak of a sheep herd in Nongan, northeast of China, the novel orf virus (ORFV) strain NA1/11 was successfully isolated. Western blot analysis indicated that the NA1/11 strain cross reacts with monoclonal antibody A3 and infected sheep ORFV antiserum. The purified virions revealed the typical ovoid shape when observed by atomic force microscopy. To determine the genetic characteristics of the NA1/11 strain, the sequences of ORFV011 (B2L), ORFV059 (F1L), ORFV109, ORFV110 and ORFv132 (VEGF) genes were amplified and compared with reference parapoxvirus strains. Non-metric multidimensional scaling (nMDS) was performed to analyze the nucleotide similarities between different ORFV strains. CONCLUSIONS Phylogenetic analysis based on ORFV 011 nucleotide sequences showed that the NA1/11strain was closely related to Xinjiang and Gansu strains. ORFV110 and ORFV132 genes are highly variable. The results revealed that precise phylogenetic analysis might provide evidence for genetic variation and movement of circulating ORFV strains in Northeast China. In addition, nMDS analysis showed that geographic isolation and animal host are likely major factors resulting in genetic differences between ORFV strains.
Collapse
Affiliation(s)
- Wei Li
- Institute of Antibody Engineering, School of Biotechnology, Southern Medical University, N, Guangzhou Avenue 1838, Guangzhou 510515, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Musser JMB, Waldron DF, Taylor CA. Evaluation of homologous and heterologous protection induced by a virulent field strain of orf virus and an orf vaccine in goats. Am J Vet Res 2012; 73:86-90. [PMID: 22204292 DOI: 10.2460/ajvr.73.1.86] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate cross protection provided by administration of contagious ecthyma vaccines against strains of orf virus in goats. ANIMALS 126 Boer-Spanish crossbred goats (3 to 20 days old). PROCEDURES 85 goats were vaccinated with a goat-derived contagious ecthyma vaccine. Of these, 41 were challenge exposed with the virus strain for the contagious ecthyma vaccine, 40 were challenge exposed with a more virulent field strain of orf virus, and 4 were lost to predation or died. Another 41 goats were vaccinated with a vaccine produced from a more virulent field strain of orf virus; of these, 18 were challenge exposed with the virus strain of the goat-derived contagious ecthyma vaccine, 18 were challenge exposed with the more virulent field strain of orf virus, and 5 were lost to predation or died. RESULTS Vaccination with the goat-derived contagious ecthyma vaccine did not significantly reduce the number of goats with lesions or lesion severity caused by challenge exposure with the more virulent field strain of orf virus. Vaccination with the vaccine produced from the more virulent field strain of orf virus significantly reduced the number of goats with lesions attributable to challenge exposure with the virus strain of the goat-derived contagious ecthyma vaccine, but it failed to significantly reduce lesion severity. CONCLUSIONS AND CLINICAL RELEVANCE Vaccination did not result in cross protection for the 2 strains of orf virus. This may have been attributable to antigenic differences and may be a factor in outbreaks of contagious ecthyma in vaccinated goats.
Collapse
Affiliation(s)
- Jeffrey M B Musser
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA.
| | | | | |
Collapse
|
32
|
Identification and characterization of monoclonal antibodies against the ORFV059 protein encoded by Orf virus. Virus Genes 2012; 44:429-40. [PMID: 22237464 DOI: 10.1007/s11262-011-0710-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 12/26/2011] [Indexed: 12/24/2022]
Abstract
Recent outbreaks of orf in China have been attributed to a novel strain of Orf virus (ORFV) designated ORFV-Jilin. Currently, monoclonal antibodies (Mabs) have not yet been developed against this specific pathogen even though such entities could have potential applications regarding the diagnosis and characterization of ORFV-Jilin. Therefore, the current study was undertaken to generate Mab against the immunodominant ORFV059 protein of this virus. For this purpose, the ORFV-Jilin ORFV059 protein was expressed in Escherichia coli and subsequently used as an antigen to immunize mice and for the initial screening of hybridomas prepared from the mice for their ability to produce anti-ORFV059 protein Mabs via an indirect ELISA. Ten, positive hybridomas were identified in this manner and verified based on the ability of their released Mab to react specifically with both naturally and artificially expressed ORFV059 protein in Western blots. The two hybridomas with the greatest propensity to secrete Mab were subcloned three times before being introduced intraperitoneally into mice. Afterwards, both Mab were separately purified from the mice's ascetic fluids and found to successfully recognize the ORFV-Jilin ORFV059 protein in a variety of immunological assays. Thus, the widespread utility of these Mab as a diagnostic core reagent should prove invaluable for further investigations regarding the mechanisms of orf pathogenesis and the control of this disease. In this regard, it should be noted that Mab A3 was used to confirm the predicted late expression of the ORFV-Jilin ORFV059 protein during virus replication.
Collapse
|
33
|
Hautaniemi M, Ueda N, Tuimala J, Mercer AA, Lahdenperä J, McInnes CJ. The genome of pseudocowpoxvirus: comparison of a reindeer isolate and a reference strain. J Gen Virol 2010; 91:1560-76. [PMID: 20107016 DOI: 10.1099/vir.0.018374-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Parapoxviruses (PPV), of the family Poxviridae, cause a pustular cutaneous disease in sheep and goats (orf virus, ORFV) and cattle (pseudocowpoxvirus, PCPV and bovine papular stomatitis virus, BPSV). Here, we present the first genomic sequence of a reference strain of PCPV (VR634) along with the genomic sequence of a PPV (F00.120R) isolated in Finland from reindeer (Rangifer tarandus tarandus). The F00.120R and VR634 genomes are 135 and 145 kb in length and contain 131 and 134 putative genes, respectively, with their genome organization being similar to that of other PPVs. The predicted proteins of F00.120R and VR634 have an average amino acid sequence identity of over 95%, whereas they share only 88 and 73% amino acid identity with the ORFV and BPSV proteomes, respectively. The most notable differences were found near the genome termini. F00.120R lacks six and VR634 lacks three genes seen near the right terminus of other PPVs. Four genes at the left end of F00.120R and one in the middle of both genomes appear to be fragmented paralogues of other genes within the genome. VR634 has larger than expected inverted terminal repeats possibly as a result of genomic rearrangements. The high G+C content (64%) of these two viruses along with amino acid sequence comparisons and whole genome phylogenetic analyses confirm the classification of PCPV as a separate species within the genus Parapoxvirus and verify that the virus responsible for an outbreak of contagious stomatitis in reindeer over the winter of 1999-2000 can be classified as PCPV.
Collapse
Affiliation(s)
- Maria Hautaniemi
- Finnish Food Safety Authority Evira, Research Department/Veterinary Virology, Mustialankatu 3, FI-00790, Helsinki, Finland.
| | | | | | | | | | | |
Collapse
|
34
|
Inoshima Y, Ishiguro N. Molecular and biological characterization of vascular endothelial growth factor of parapoxviruses isolated from wild Japanese serows (Capricornis crispus). Vet Microbiol 2010; 140:63-71. [DOI: 10.1016/j.vetmic.2009.07.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 07/18/2009] [Accepted: 07/31/2009] [Indexed: 11/30/2022]
|
35
|
Virus-encoded homologs of cellular interleukin-10 and their control of host immune function. J Virol 2009; 83:9618-29. [PMID: 19640997 DOI: 10.1128/jvi.01098-09] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
36
|
Effect of local viral transfer of interleukin 10 gene on a rabbit arthritis model induced by interleukin 1β. Chin Med J (Engl) 2008. [DOI: 10.1097/00029330-200803010-00013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|